Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Blood ; 125(3): 570-80, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25352130

ABSTRACT

The common γ chain (CD132) is a subunit of the interleukin (IL) receptors for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Because levels of several of these cytokines were shown to be increased in the serum of patients developing acute and chronic graft-versus-host disease (GVHD), we reasoned that inhibition of CD132 could have a profound effect on GVHD. We observed that anti-CD132 monoclonal antibody (mAb) reduced acute GVHD potently with respect to survival, production of tumor necrosis factor, interferon-γ, and IL-6, and GVHD histopathology. Anti-CD132 mAb afforded protection from GVHD partly via inhibition of granzyme B production in CD8 T cells, whereas exposure of CD8 T cells to IL-2, IL-7, IL-15, and IL-21 increased granzyme B production. Also, T cells exposed to anti-CD132 mAb displayed a more naive phenotype in microarray-based analyses and showed reduced Janus kinase 3 (JAK3) phosphorylation upon activation. Consistent with a role of JAK3 in GVHD, Jak3(-/-) T cells caused less severe GVHD. Additionally, anti-CD132 mAb treatment of established chronic GVHD reversed liver and lung fibrosis, and pulmonary dysfunction characteristic of bronchiolitis obliterans. We conclude that acute GVHD and chronic GVHD, caused by T cells activated by common γ-chain cytokines, each represent therapeutic targets for anti-CD132 mAb immunomodulation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Marrow Transplantation/adverse effects , Cytokines/metabolism , Graft vs Host Disease/prevention & control , Interleukin Receptor Common gamma Subunit/antagonists & inhibitors , Acute Disease , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Chronic Disease , Female , Flow Cytometry , Fluorescent Antibody Technique , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Humans , Janus Kinase 3/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
J Pharmacol Exp Ther ; 348(1): 165-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24218541

ABSTRACT

A critical piece in the translation of preclinical studies to clinical trials is the determination of dosing regimens that allow maximum therapeutic benefit with minimum toxicity. The preclinical pharmacokinetic (PK)/pharmacodynamic (PD) profile of tofacitinib, an oral Janus kinase (JAK) inhibitor, in a mouse collagen-induced arthritis (mCIA) model was compared with clinical PK/PD data from patients with rheumatoid arthritis (RA). Preclinical evaluations included target modulation and PK/PD modeling based on continuous subcutaneous infusion or oral once- or twice-daily (BID) dosing paradigms in mice. The human PK/PD profile was obtained from pooled data from four phase 2 studies in patients with RA, and maximal effect models were used to evaluate efficacy after 12 weeks of tofacitinib treatment (1-15 mg BID). In mCIA, the main driver of efficacy was inhibition of cytokine receptor signaling mediated by JAK1 heterodimers, but not JAK2 homodimers, and continuous daily inhibition was not required to maintain efficacy. Projected efficacy could be predicted from total daily exposure irrespective of the oral dosing paradigm, with a total steady-state plasma concentration achieving 50% of the maximal response (Cave50) of ~100 nM. Tofacitinib potency (ED50) in clinical studies was ~3.5 mg BID (90% confidence interval: 2.3, 5.5) or total Cave50 of ~40 nM, derived using Disease Activity Scores from patients with RA. The collective clinical and preclinical data indicated the importance of Cave as a driver of efficacy, rather than maximum or minimum plasma concentration (Cmax or Cmin), where Cave50 values were within ~2-fold of each other.


Subject(s)
Arthritis, Experimental/drug therapy , Janus Kinase 1/antagonists & inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Animals , Arthritis, Experimental/enzymology , Double-Blind Method , Drug Evaluation, Preclinical/methods , Humans , Janus Kinase 1/metabolism , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/physiology , Male , Mice , Mice, Inbred DBA , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use
3.
J Membr Biol ; 247(8): 713-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24928228

ABSTRACT

Janus kinase 3 (JAK3) contributes to cytokine receptor signaling, confers cell survival and stimulates cell proliferation. The gain of function mutation JAK3(A572V) is found in acute megakaryoplastic leukemia. Replacement of ATP coordinating lysine by alanine yields inactive JAK3(K855A). Most recent observations revealed the capacity of JAK3 to regulate ion transport. This study thus explored whether JAK3 regulates glutamate transporters EAAT1-4, carriers accomplishing transport of glutamate and aspartate in a variety of cells including intestinal cells, renal cells, glial cells, and neurons. To this end, EAAT1, 2, 3, or 4 were expressed in Xenopus oocytes with or without additional expression of mouse wild-type JAK3, constitutively active JAK3(A568V) or inactive JAK3(K851A), and electrogenic glutamate transport was determined by dual electrode voltage clamp. Moreover, Ussing chamber was employed to determine electrogenic glutamate transport in intestine from mice lacking functional JAK3 (jak3(-/-)) and from corresponding wild-type mice (jak3(+/+)). As a result, in EAAT1, 2, 3, or 4 expressing oocytes, but not in oocytes injected with water, addition of glutamate to extracellular bath generated an inward current (Ig), which was significantly increased following coexpression of JAK3. Ig in oocytes expressing EAAT3 was further increased by JAK3(A568V) but not by JAK3(K851A). Ig in EAAT3 + JAK3 expressing oocytes was significantly decreased by JAK3 inhibitor WHI-P154 (22 µM). Kinetic analysis revealed that JAK3 increased maximal Ig and significantly reduced the glutamate concentration required for half maximal Ig (Km). Intestinal electrogenic glutamate transport was significantly lower in jak3(-/-) than in jak3(+/+) mice. In conclusion, JAK3 is a powerful regulator of excitatory amino acid transporter isoforms.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Glutamic Acid/metabolism , Janus Kinase 3/physiology , Oocytes/metabolism , Signal Transduction , Animals , Cell Proliferation , Mice , Mice, Knockout , Patch-Clamp Techniques , Up-Regulation , Xenopus laevis
4.
J Neuroinflammation ; 9: 170, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22788969

ABSTRACT

BACKGROUND: Secreted phospholipase A2-IIA (sPLA2-IIA) is an inducible enzyme released under several inflammatory conditions. It has been shown that sPLA2-IIA is released from rat brain astrocytes after inflammatory stimulus, and lipopolysaccharide (LPS) and nitric oxide (NO) have been implicated in regulation of this release. Here, brain microvascular endothelial cells (BMVECs) were treated with LPS to uncover whether sPLA2-IIA was released, whether nitric oxide regulated this release, and any related signal mechanisms. METHODS: Supernatants were collected from primary cultures of BMVECs. The release of sPLA2-IIA, and the expression of inducible nitric oxide synthase (iNOS), phospho-JAK3, phospho-STAT1, total JAK3 and STAT1, ß-actin, and bovine serum albumin (BSA) were analyzed by Western blot or ELISA. NO production was calculated by the Griess reaction. sPLA2 enzyme activity was measured with a fluorometric assay. Specific inhibitors of NO (L-NAME and aminoguanidine, AG), JAK3 (WHI-P154,WHI), STAT1 (fludarabine, Flu), and STAT1 siRNA were used to determine the involvement of these molecules in the LPS-induced release of sPLA2-IIA from BMVECs. Nuclear STAT1 activation was tested with the EMSA method. The monolayer permeability of BMVECs was measured with a diffusion assay using biotinylated BSA. RESULTS: Treatment of BMVECs with LPS increased the release of sPLA2-IIA and nitrite into the cell culture medium up to 24 h. Pretreatment with an NO donor, sodium nitroprusside, decreased LPS-induced sPLA2-IIA release and sPLA2 enzyme activity, and enhanced the expression of iNOS and nitrite generation after LPS treatment. Pretreatment with L-NAME, AG, WHI-P154, or Flu notably reduced the expression of iNOS and nitrite, but increased sPLA2-IIA protein levels and sPLA2 enzyme activity. In addition, pretreatment of the cells with STAT1 siRNA inhibited the phosphorylation of STAT1, iNOS expression, and nitrite production, and enhanced the release of sPLA2-IIA. Pretreatment with the specific inhibitors of NOS, JAK2, and STAT3 decreased the permeability of BMVECs. In contrast, inhibition of sPLA2-IIA release increased cell permeability. These results suggest that sPLA2-IIA expression is regulated by the NO-JAK3-STAT1 pathway. Importantly, sPLA2-IIA augmentation could protect the LPS-induced permeability of BMVECs. CONCLUSION: Our results demonstrate the important action of sPLA2-IIA in the permeability of microvascular endothelial cells during brain inflammatory events. The sPLA2 and NO pathways can be potential targets for the management of brain MVEC injuries and related inflammation.


Subject(s)
Brain Injuries/metabolism , Endothelium, Vascular/metabolism , Group II Phospholipases A2/metabolism , Janus Kinase 3/physiology , Microcirculation/physiology , STAT1 Transcription Factor/physiology , Animals , Brain Injuries/enzymology , Brain Injuries/pathology , Cell Membrane Permeability , Cells, Cultured , Endothelium, Vascular/enzymology , Group II Phospholipases A2/biosynthesis , Group II Phospholipases A2/genetics , Lipopolysaccharides/pharmacology , Microcirculation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
5.
Blood ; 113(12): 2746-54, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19139084

ABSTRACT

The tyrosine kinase JAK3 plays a well-established role during normal lymphocyte development and is constitutively phosphorylated in several lymphoid malignancies. However, its contribution to lymphomagenesis remains elusive. In this study, we used the newly identified activating JAK3A572V mutation to elucidate the effect of constitutive JAK3 signaling on murine lymphopoiesis. In a bone marrow transplantation model, JAK3A572V induces an aggressive, fatal, and transplantable lymphoproliferative disorder characterized by the expansion of CD8(+)TCRalphabeta(+)CD44(+)CD122(+)Ly-6C(+) T cells that closely resemble an effector/memory T-cell subtype. Compared with wild-type counterparts, these cells show increased proliferative capacities in response to polyclonal stimulation, enhanced survival rates with elevated expression of Bcl-2, and increased production of interferon-gamma (IFNgamma) and tumor necrosis factor-alpha (TNFalpha), correlating with enhanced cytotoxic abilities against allogeneic target cells. Of interest, the JAK3A572V disease is epidermotropic and produces intraepidermal microabscesses. Taken together, these clinical features are reminiscent of those observed in an uncommon but aggressive subset of CD8(+) human cutaneous T-cell lymphomas (CTCLs). However, we also observed a CD4(+) CTCL-like phenotype when cells are transplanted in an MHC-I-deficient background. These data demonstrate that constitutive JAK3 activation disrupts T-cell homeostasis and induces lymphoproliferative diseases in mice.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Janus Kinase 3/physiology , Lymphopoiesis/physiology , Lymphoproliferative Disorders/etiology , Point Mutation , Recombinant Fusion Proteins/physiology , T-Lymphocyte Subsets/pathology , Animals , Antigens, Ly/analysis , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/chemistry , Enzyme Induction , Humans , Hyaluronan Receptors/analysis , Interleukin-2 Receptor beta Subunit/analysis , Janus Kinase 3/biosynthesis , Janus Kinase 3/genetics , Lymphoma, T-Cell, Cutaneous/pathology , Lymphopoiesis/genetics , Lymphoproliferative Disorders/enzymology , Lymphoproliferative Disorders/pathology , Mice , Mice, Inbred C57BL , Radiation Chimera , Receptors, Antigen, T-Cell, alpha-beta/analysis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Skin/pathology , T-Lymphocyte Subsets/chemistry
6.
J Immunol ; 183(3): 2183-92, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19596999

ABSTRACT

JAK3, a member of the Janus kinase family, is predominantly expressed in hemopoietic cells and binds specifically to the common gamma chain of a subfamily of cytokine receptors that includes IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Previous studies suggest that this tyrosine kinase plays key roles in mediating T cell functions, and inhibition of JAK3 has been shown to prevent graft rejection and decrease the severity of arthritis in rodent models. However, the functions of JAK3 in the development of skin immune responses and diseases such as psoriasis have not been determined. CD18 mutant PL/J mice develop spontaneous T cell-dependent psoriasiform skin disease with several similarities to human psoriasis. In this study, we treated mice with established skin disease with R348, a small molecule inhibitor of JAK3, and observed a marked attenuation of skin lesions following 6 wk of treatment. Histological analyses revealed major reductions of both epidermal and dermal lesion severity scores in R348-treated CD18-deficient PL/J mice compared with vehicle controls, which was associated with decreased CD4(+) T cell infiltration. In addition, systemic levels of IL-17, IL-22, IL-23, and TNF-alpha were significantly lower in mice receiving the compound, and T cells isolated from R348-treated mice also showed reduced phosphorylation of Stat5 after stimulation with IL-2. These findings suggest that small-molecule inhibitors of JAK3 may be useful in the treatment of inflammatory skin diseases such as psoriasis and strongly implicate JAK signaling events as important in the pathogenesis of this disease.


Subject(s)
CD18 Antigens/genetics , Inflammation/drug therapy , Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Psoriasis/drug therapy , Animals , CD4-Positive T-Lymphocytes/physiology , Chemotaxis , Cytokines/analysis , Janus Kinase 3/immunology , Janus Kinase 3/physiology , Mice , Mice, Mutant Strains , Protein Kinase Inhibitors/pharmacology , Psoriasis/pathology , STAT5 Transcription Factor/metabolism , Treatment Outcome
7.
J Immunol ; 183(7): 4493-501, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734221

ABSTRACT

T cell proliferation following activation is an essential aspect of the adaptive immune response. Multiple factors, such as TCR signaling, costimulation, and signals from cytokines, each contribute to determine the magnitude of T cell expansion. In this report, we examine in detail the role of Jak3/common gamma-chain-dependent cytokines in promoting cell cycle progression and proliferation of naive T cells. Using naive CD4+ T cells from Jak3-deficient mice and wild-type CD4+ T cells treated with a small molecule inhibitor of Jak3, we find that these cytokine signals are not required for proliferation; instead, they are important for the survival of activated T cells. In addition, we show that the percentage of cells entering the cell cycle and the percentage of cells in each round of cell division are comparable between Jak3-deficent and wild-type T cells. Furthermore, cell cycle progression and the regulated expression of key cell cycle proteins are independent of Jak3/common gamma-chain cytokine signals. These findings hold true over a wide range of TCR signal strengths. However, when CD28 costimulatory signals, but not TCR signals, are limiting, Jak3-dependent cytokine signals become necessary for the proliferation of naive T cells. Because CD28 signaling has been found to be dispensable for autoreactive T cell responses, these data suggest the potential for interfering with autoimmune T cell responses by inhibition of Jak3 signaling.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Cycle/immunology , Interleukin Receptor Common gamma Subunit/physiology , Janus Kinase 3/physiology , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Cycle/genetics , Cell Proliferation/drug effects , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Neurochem Res ; 34(6): 1101-12, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19009346

ABSTRACT

Nerve growth factor (NGF)-mediated activation of mitogen-activated protein kinases (MAPK) is critical for differentiation and apoptosis of PC12 cells. Since NGF employs stress-activated c-Jun N-terminal kinase (JNK) to regulate both programmed cell death and neurite outgrowth of PC12 cells, we examined NGF-regulated JNK activity and the role of G(i/o) proteins. Induction of JNK phosphorylation by NGF occurred in a time- and dose-dependent manner and was partially inhibited by pertussis toxin (PTX). To discern the participation of various signaling intermediates, PC12 cells were treated with specific inhibitors prior to NGF challenge. NGF-elevated JNK activity was abolished by inhibitors of JNK, p38 MAPK, Src, JAK3 and MEK1/2. NGF-dependent JNK phosphorylation became insensitive to PTX treatment upon transient expressions of Galpha(z) or the PTX-resistant mutants of Galpha(i1-3) and Galpha(oA). Collectively, these studies indicate that NGF-dependent JNK activity may be mediated via G(i1-3) proteins, JAK3, Src, p38 MAPK and the MEK/ERK cascade.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/physiology , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/physiology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/physiology , Mutation , Neurons/metabolism , PC12 Cells , Pertussis Toxin/pharmacology , Phosphorylation , Rats , Receptor, trkA/physiology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/physiology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/physiology
9.
Clin Dysmorphol ; 28(2): 57-62, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30614825

ABSTRACT

Stüve-Wiedemann syndrome (OMIM #601559) is a rare, autosomal recessive disorder characterized by skeletal dysplasia, consecutive infections, feeding difficulties and autonomic dysregulation. We present an Afro-Caribbean family with two siblings diagnosed with Stüve-Wiedemann syndrome. The underlying loss-of-function mutation in the leukemia inhibitory factor receptor gene is thought to impair proper functioning of the JAK/STAT 3 pathway. As this affects normal functioning of T-helper cells, these patients are prone to infections with uncommon pathogens as illustrated by this case.


Subject(s)
Exostoses, Multiple Hereditary/physiopathology , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Osteochondrodysplasias/physiopathology , Abnormalities, Multiple/genetics , Adult , Family , Female , Humans , Infant, Newborn , Janus Kinase 3/physiology , Janus Kinases/physiology , Leukemia Inhibitory Factor Receptor alpha Subunit/physiology , Male , Mutation , Pedigree , STAT3 Transcription Factor/physiology , Siblings , Syndrome
10.
Int Immunopharmacol ; 6(3): 433-44, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16428079

ABSTRACT

Down-regulation of cell-mediated immune functions occurring at late stages of cancer may be related to the thymic involution since thymus is the major site of T cell maturation, proliferation, and differentiation. We found that in Ehrlich's ascites carcinoma (EAC)-bearing mice there was profound depletion of CD4+ and CD8+ cells in peripheral blood with severely damaged thymus on 21st day of tumor inoculation. However, treatment with black tea at an antitumor dose of 2.5% significantly reduced such depletion and protected the thymus considerably from tumor onslaught. A search for the underlying mechanism revealed EAC-induced IL-7Ralpha down-regulation, inhibition of JAK3 and STAT5 phosphorylation, and decrease in Bcl-2/Bax ratio in thymocytes that finally led to thymocyte apoptosis in one hand and T cell maturation block on the other. Interestingly, black tea treatment prevented IL-7Ralpha down-regulation and protected the signaling cascade through JAK-STAT thereby inhibiting tumor-induced thymic apoptosis and ensuring proper functioning of this organ in tumor-bearing host.


Subject(s)
Carcinoma, Ehrlich Tumor/pathology , Carcinoma, Ehrlich Tumor/prevention & control , Janus Kinase 3/antagonists & inhibitors , MAP Kinase Signaling System/immunology , Receptors, Interleukin-7/antagonists & inhibitors , STAT5 Transcription Factor/antagonists & inhibitors , Tea , Thymus Gland/pathology , Animals , Apoptosis/physiology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Camellia sinensis , Carcinoma, Ehrlich Tumor/enzymology , Carcinoma, Ehrlich Tumor/immunology , Cell Line, Tumor , Cells, Cultured , Janus Kinase 3/physiology , Leukocyte Count , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Lymphocyte Count , Mice , Phosphorylation , Plant Extracts/therapeutic use , Receptors, Interleukin-7/physiology , STAT5 Transcription Factor/physiology , Tea/chemistry , Thymus Gland/drug effects , Thymus Gland/enzymology , Thymus Gland/immunology
11.
PLoS One ; 9(2): e88014, 2014.
Article in English | MEDLINE | ID: mdl-24498424

ABSTRACT

We have previously shown that Jak3 is involved in the signaling pathways of CCR7, CCR9 and CXCR4 in murine T lymphocytes and that Jak3⁻/⁻ lymphocytes display an intrinsic defect in homing to peripheral lymph nodes. However, the molecular mechanism underlying the defective migration observed in Jak3⁻/⁻ lymphocytes remains elusive. Here, it is demonstrated for the first time, that Jak3 is required for the actin cytoskeleton reorganization in T lymphocytes responding to chemokines. It was found that Jak3 regulates actin polymerization by controlling cofilin inactivation in response to CCL21 and CXCL12. Interestingly, cofilin inactivation was not precluded in PTX- treated cells despite their impaired actin polymerization. Additionally, Jak3 was required for small GTPases Rac1 and RhoA activation, which are indispensable for acquisition of the migratory cell phenotype and the generation of a functional leading edge and uropod, respectively. This defect correlates with data obtained by time-lapse video-microscopy showing an incompetent uropod formation and impaired motility in Jak3-pharmacologically inhibited T lymphocytes. Our data support a new model in which Jak3 and heterotrimeric G proteins can use independent, but complementary, signaling pathways to regulate actin cytoskeleton dynamics during cell migration in response to chemokines.


Subject(s)
Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Chemokine CCL21/pharmacology , Chemokine CXCL12/pharmacology , Janus Kinase 3/physiology , rac GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/immunology , Animals , Cell Movement/drug effects , Chemotaxis, Leukocyte/drug effects , Female , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Pertussis Toxin/pharmacology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time-Lapse Imaging
12.
AIDS Res Hum Retroviruses ; 29(5): 784-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23298197

ABSTRACT

The subcellular localization of JAK3 was examined by quantitative image analysis. For the first time, JAK3 was found to be located in the nuclei of primary CD4 lymphocytes. A comparable quantity of JAK3 was recovered in CD4 lymphocytes from healthy donors and HIV-infected patients. By contrast, far more phosphorylated JAK3 (pJAK3) was found in the nuclei of CD4 lymphocytes from HIV-infected patients than from healthy donors. The correlation detected between the quantity of pJAK3 in the nuclei of CD4 lymphocytes and the increase in HLA-DR at their surface suggests that pJAK3 may play a role in the deleterious immune activation characterizing HIV-infected patients.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Cell Nucleus/physiology , HIV Infections/immunology , Janus Kinase 3/physiology , Lymphocyte Activation/physiology , Humans , Lymphocyte Activation/immunology
13.
Cancer Lett ; 333(1): 47-55, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23348693

ABSTRACT

NK cell neoplasms are lymphoid malignancies with an aggressive clinical course. In the present study, we analyzed gene expression profiling of NK cell neoplasms and attempted to identify important molecular pathways and new effective drugs. Pathway analysis of gene expression profiles suggested the important roles of the JAK-STAT pathway, NF-κB pathway or Wnt pathways in NK cell neoplasms. Notably, western blot analysis revealed that STAT3 was expressed and phosphorylated at a higher level in NK cell lines than in normal NK cells or other cell lines. These findings indicate the occurrence of JAK-STAT activation in NK cell neoplasms. Connectivity Map (CMAP) analysis of gene expression profiles identified candidate drugs against NK cell neoplasms. Among the drugs suggested by CMAP analysis, we focused on puromycin, phenoxybenzamine, LY294002, wortmannin, vorinostat and trichostatin A because they exhibited high enrichment scores. We added these drugs to NK cell lines and other cell lines. Among the drugs, vorinostat suppressed NK cell line proliferation at a significantly lower concentration compared to other cell lines. Suppression of the JAK-STAT pathway appeared to contribute to this effect. Vorinostat may be a good candidate for use in the therapy against NK cell neoplasms.


Subject(s)
Antineoplastic Agents/pharmacology , Hydroxamic Acids/pharmacology , Killer Cells, Natural/pathology , Lymphoma/drug therapy , Transcriptome , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Janus Kinase 3/physiology , Lymphoma/immunology , Lymphoma/pathology , STAT3 Transcription Factor/physiology , Vorinostat
14.
J Cancer Res Clin Oncol ; 139(9): 1551-62, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23877657

ABSTRACT

PURPOSE: Cholangiocarcinoma (CCA) or cancer of the biliary tract is heterogeneous; however, chronic inflammatory-related features are unique in CCA. Moreover, the genes involved in proteasome functions are evidently increased in CCA. Hence, CCA might be vulnerable to endoplasmic reticulum (ER) stressors, particularly a proteasome inhibitor. Therefore, bortezomib (BTZ), a specific 26S proteasome inhibitor, was selected, and its antitumor effects against CCA were investigated. METHODS: Liver fluke-associated CCA cell lines were used. Cell proliferation and apoptosis detection were determined by a tetrazolium-based assay, caspase detection and annexin V binding assay. The accumulations of proteasome substrates, the inductions of ER stress and unfolded protein response (UPR) proteins were demonstrated by western blot and reporter systems. The in vivo anti-proliferative effect was accessed in a subcutaneous transplantation mouse model. RESULTS: BTZ inhibited CCA proliferation and induced caspase-dependent apoptosis, independently of the NF-κB pathway. Inhibition of protein degradation by BTZ led to the induction of UPR; induction of XBP1 splicing, ATF6 proteolysis and nuclear ATF4 as well as BiP and CHOP expressions were evident. Nevertheless, ER stress-induced UPR was overwhelming, leading to the activation of apoptosis demonstrated by proteolytic cleavages of ER-related caspase 4 and 12 as well as classical caspase 8, 9 and 3. The growth inhibitory effect of BTZ was supported by an in vivo model. CONCLUSION: BTZ treatment could be a promising therapeutic approach for CCA treatment.


Subject(s)
Apoptosis/drug effects , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Boronic Acids/pharmacology , Cholangiocarcinoma/pathology , Endoplasmic Reticulum Stress/drug effects , Janus Kinase 3/physiology , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/drug effects , Bile Ducts, Intrahepatic/metabolism , Blotting, Western , Bortezomib , Cell Proliferation/drug effects , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Humans , Luciferases/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , NF-kappa B/metabolism , Transcription Factor CHOP/metabolism , Tumor Cells, Cultured , Unfolded Protein Response/drug effects
15.
J Invest Dermatol ; 131(6): 1331-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21346774

ABSTRACT

IL-17 is a proinflammatory cytokine that is crucial for the host's protection against a range of extracellular pathogens. However, inappropriately regulated expression of IL-17 is associated with the development of inflammatory diseases and cancer. In cutaneous T-cell lymphoma (CTCL), malignant T cells gradually accumulate in skin lesions characterized by massive chronic inflammation, suggesting that IL-17 could be involved in the pathogenesis. In this study we show that IL-17 protein is present in 10 of 13 examined skin lesions but not in sera from 28 CTCL patients. Importantly, IL-17 expression is primarily observed in atypical lymphocytes with characteristic neoplastic cell morphology. In accordance, malignant T-cell lines from CTCL patients produce IL-17 and the synthesis is selectively increased by IL-2 receptor ß chain cytokines. Small-molecule inhibitors or small interfering RNA against Jak3 and signal transducer and activator of transcription 3 (Stat3) reduce the production of IL-17, showing that the Jak3/Stat3 pathway promotes the expression of the cytokine. In summary, our findings indicate that the malignant T cells in CTCL lesions express IL-17 and that this expression is promoted by the Jak3/Stat3 pathway.


Subject(s)
Interleukin-17/physiology , Janus Kinase 3/physiology , Lymphoma, T-Cell, Cutaneous/immunology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Skin Neoplasms/immunology , Cell Line, Tumor , Humans , Interleukin-17/analysis , Lymphoma, T-Cell, Cutaneous/etiology , Skin Neoplasms/etiology , T-Lymphocytes/immunology
16.
Neurotox Res ; 19(3): 484-95, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20717763

ABSTRACT

We have previously shown that interleukin-6 (IL-6)-protected neurons against the suppression of neuronal vitality and overload of intracellular Ca(2+) induced by glutamate or N-methyl-D: -aspartate (NMDA). Herein we provide further evidence for IL-6 neuroprotection against NMDA-induced apoptosis and explore the signal-transduction mechanisms underlying the anti-apoptotic action of IL-6. Cerebellar granule neurons (CGNs) from postnatal 8-day infant rats were chronically exposed to IL-6 (40 or 120 ng/ml) for 8 days, and stimulated with NMDA (100 µM) for 30 min. To observe the signaling pathways, we employed AG490 (5 or 10 µM), an inhibitor of Janus kinases (JAKs), or LY294002 (5 or 10 µM), an inhibitor of phosphatidylinositol 3-kinase (PI3K), to pretreat the CGNS together with IL-6. The levels of phosphorylation for the downstream effectors of JAKs and PI3K, i.e., phosphorylated STAT3 and Akt, were quantified by Western blot assay. In the cultured CGNs with various drug exposures, the expressions of Bcl-2, Bax, and caspase-3 were measured by real-time PCR and Western blot, and the percentage of apoptotic nuclei was tested by Hoechst 33342 staining. After the CGNs were chronically exposed to IL-6, NMDA stimulation led to an increase in the expression of Bcl-2 mRNA and a decrease in the expression of Bax and caspase-3 mRNAs and proteins when compared with those neurons lacking IL-6 exposure. IL-6 pretreatment of the neurons without NMDA stimulation concentration-dependently enhanced the expressions of Bcl-2 mRNA and protein while attenuating the expressions of Bax and caspase-3 mRNAs and proteins in comparison with control lacking any treatment. Furthermore, IL-6 prevented the increase in the percentage of apoptotic neurons induced by NMDA. The combined pretreatment of the CGNs with AG490 and IL-6 or with LY294002 and IL-6 reduced these anti-apoptotic effects of IL-6. Neither AG490 nor LY294002 exposure alone altered the expressions of Bcl-2, Bax, and cleaved caspase-3 proteins. IL-6 up-regulated the levels of phosphorylated STAT3 and Akt, and this was blocked by AG490 and LY294002, respectively. These results suggest that IL-6 protects neurons against NMDA-induced apoptosis, and that the IL-6 neuroprotection is jointly mediated by JAK-STAT3 and PI3K-Akt signaling pathways.


Subject(s)
Apoptosis/physiology , Cytokine Receptor gp130/physiology , Interleukin-6/physiology , N-Methylaspartate/toxicity , Neuroprotective Agents/pharmacology , Signal Transduction/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Janus Kinase 3/physiology , N-Methylaspartate/antagonists & inhibitors , Neurons/metabolism , Neurons/pathology , Neurons/physiology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/physiology , Signal Transduction/drug effects
17.
PLoS One ; 4(9): e7066, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19759904

ABSTRACT

BACKGROUND: CCR7-mediated signalling is important for dendritic cell maturation and homing to the lymph nodes. We have previously demonstrated that Jak3 participates in the signalling pathway of CCR7 in T lymphocytes. METHODOLOGY AND PRINCIPAL FINDINGS: Here, we used Jak3(-/-) mice to analyze the role of Jak3 in CCR7-mediated dendritic cells migration and function. First, we found no differences in the generation of DCs from Jak3(-/-) bone marrow progenitors, when compared to wild type cells. However, phenotypic analysis of the bone marrow derived DCs obtained from Jak3(-/-) mice showed reduced expression of co-stimulatory molecules compared to wild type (Jak3(+/+)). In addition, when we analyzed the migration of Jak3(-/-) and Jak3(+/+) mature DCs in response to CCL19 and CCL21 chemokines, we found that the absence of Jak3 results in impaired chemotactic responses both in vitro and in vivo. Moreover, lymphocyte proliferation and contact hypersensitivity experiments showed that DC-mediated T lymphocyte activation is reduced in the absence of Jak3. CONCLUSION/SIGNIFICANCE: Altogether, our data provide strong evidence that Jak3 is important for DC maturation, migration and function, through a CCR7-mediated signalling pathway.


Subject(s)
Dendritic Cells/cytology , Gene Expression Regulation , Janus Kinase 3/physiology , Receptors, CCR7/physiology , Animals , Bone Marrow Cells/cytology , Cell Movement , Cell Proliferation , Chemotaxis , Lymphocyte Activation , Lymphocytes/cytology , Mice , Mice, Transgenic , Models, Biological , T-Lymphocytes/metabolism
18.
J Thorac Oncol ; 4(4): 448-54, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19240653

ABSTRACT

BACKGROUND: With the emergence of Src inhibitors in clinical trials, improved knowledge of the molecular responses of cancer cells to these agents is warranted. This will facilitate the development of tests to identify patients who may benefit from these agents, allow drug activity to be monitored and rationalize the combination of these agents with other treatment modalities. METHODS: This study evaluated the molecular and functional effects of Src inhibitor AZD0530 in human lung cancer cells, by Western blotting and reverse transcription-polymerase chain reaction, and by assays for cell viability, migration, and invasion. RESULTS: Src was activated in four of five cell lines tested and the level corresponded with the invasive potential and the histologic subtype. Clinically relevant, submicromolar concentrations of AZD0530 blocked Src and focal adhesion kinase, resulting in significant inhibition of cell migration and Matrigel invasion. Reactivation of STAT3 and up-regulation of JAK indicated a potential mechanism of resistance. AZD0530 gave a potent and sustained blockage of AKT and enhanced the sensitivity to irradiation. CONCLUSIONS: The results indicated that AZD0530, aside from being a potent inhibitor of tumor cell invasion which could translate to inhibition of disease progression in the clinic, may also lower resistance of lung cancer cells to pro-apoptotic signals.


Subject(s)
Benzodioxoles/pharmacology , Lung Neoplasms/radiotherapy , Quinazolines/pharmacology , Radiation-Sensitizing Agents/pharmacology , src-Family Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Chromones/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/physiology , Humans , Janus Kinase 3/genetics , Janus Kinase 3/physiology , Lung Neoplasms/pathology , Morpholines/pharmacology , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , STAT3 Transcription Factor/analysis , STAT3 Transcription Factor/physiology , Signal Transduction
19.
Biochem Biophys Res Commun ; 354(3): 825-9, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17266928

ABSTRACT

Binding of interleukin-2 (IL-2) to its specific receptor induces activation of two members of Jak family protein tyrosine kinases, Jak1 and Jak3. An IL-2 receptor (IL-2R)-reconstituted NIH 3T3 fibroblast cell line proliferates in response to IL-2 only when hematopoietic lineage-specific Jak3 is ectopically expressed. However, the mechanism of Jak3-dependent proliferation in the fibroblast cell line is not known. Here, I showed that Jak3 expression is dispensable for IL-2-induced activation of Jak1 and Stat proteins and expression of nuclear proto-oncogenes in the IL-2R-reconstituted fibroblast cell line. Jak3 expression markedly enhanced these IL-2-induced signaling events. In contrast, Jak3 expression was essential for induction of cyclin genes involved in the G1-S transition. These data suggest a critical role of Jak3 in IL-2 signaling in the fibroblast cell line and may provide further insight into the cell type-specific mechanism of cytokine signaling.


Subject(s)
Fibroblasts/cytology , Interleukin-2/physiology , Janus Kinase 3/physiology , Signal Transduction/physiology , Animals , Binding Sites , Cell Proliferation , Cyclins/genetics , Cyclins/metabolism , Cytokines/genetics , Cytokines/metabolism , G1 Phase/genetics , G1 Phase/physiology , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 3/genetics , Mice , NIH 3T3 Cells/cytology , S Phase/genetics , S Phase/physiology , STAT Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL